Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int J Mol Sci ; 24(21)2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37958526

ABSTRACT

Transplanted mesenchymal stromal cells (MSCs) exhibit a robust anti-inflammatory and homing capacity in response to high inflammatory signals, as observed in studies focused on rheumatic diseases that target articular cartilage (AC) health. However, AC degradation in osteoarthritis (OA) does not necessarily coincide with a highly inflammatory joint profile. Often, by the time patients seek medical attention, they already have damaged AC. In this study, we examined the therapeutic potential of a single bone marrow MSC transplant (2 × 106 cells/kgbw) through two different routes: intra-articular (MSCs-IAt) and intravenous (MSCs-IVt) in a preclinical model of low-grade inflammatory OA with an established AC degeneration. OA was induced through the destabilization of the medial meniscus (DMM) in female Wistar Kyoto rats. The animals received MSCs 9 weeks after surgery and were euthanized 4 and 12 weeks post-transplant. In vivo and ex vivo tracking of MSCs were analyzed via bioluminescence and imaging flow cytometry, respectively. Cytokine/chemokine modulation in serum and synovial fluid was measured using a multiplex panel. AC degeneration was quantified through histology, and hindlimb muscle balance was assessed with precision weighing. To our knowledge, we are the first group to show the in vivo (8 h) and ex vivo (12 h) homing of cells to the DMM-OA joint following MSCs-IVt. In the case of MSCs-IAt, the detection of cellular bioluminescence at the knee joint persisted for up to 1 week. Intriguingly, intra-articular saline injection (placebo-IAt) resulted in a worse prognosis of OA when compared to a non-invasive control (placebo-IVt) without joint injection. The systemic cytokines/chemokines profile exhibited a time-dependent variation between transplant routes, displaying a transient anti-inflammatory systemic response for both MSCs-IVt and MSCs-IAt. A single injection of MSCs, whether administered via the intra-articular or intravenous route, performed 9 weeks after DMM surgery, did not effectively inhibit AC degeneration when compared to a non-invasive control.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Osteoarthritis , Humans , Rats , Female , Animals , Menisci, Tibial/metabolism , Osteoarthritis/metabolism , Cartilage, Articular/metabolism , Anti-Inflammatory Agents/pharmacology , Injections, Intra-Articular , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cell Transplantation/methods
2.
Int J Mol Sci, v.24, n. 21, 15543, set. 2023
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-5148

ABSTRACT

Transplanted mesenchymal stromal cells (MSCs) exhibit a robust anti-inflammatory and homing capacity in response to high inflammatory signals, as observed in studies focused on rheumatic diseases that target articular cartilage (AC) health. However, AC degradation in osteoarthritis (OA) does not necessarily coincide with a highly inflammatory joint profile. Often, by the time patients seek medical attention, they already have damaged AC. In this study, we examined the therapeutic potential of a single bone marrow MSC transplant (2 × 106 cells/kgbw) through two different routes: intra-articular (MSCs-IAt) and intravenous (MSCs-IVt) in a preclinical model of low-grade inflammatory OA with an established AC degeneration. OA was induced through the destabilization of the medial meniscus (DMM) in female Wistar Kyoto rats. The animals received MSCs 9 weeks after surgery and were euthanized 4 and 12 weeks post-transplant. In vivo and ex vivo tracking of MSCs were analyzed via bioluminescence and imaging flow cytometry, respectively. Cytokine/chemokine modulation in serum and synovial fluid was measured using a multiplex panel. AC degeneration was quantified through histology, and hindlimb muscle balance was assessed with precision weighing. To our knowledge, we are the first group to show the in vivo (8 h) and ex vivo (12 h) homing of cells to the DMM–OA joint following MSCs-IVt. In the case of MSCs-IAt, the detection of cellular bioluminescence at the knee joint persisted for up to 1 week. Intriguingly, intra-articular saline injection (placebo-IAt) resulted in a worse prognosis of OA when compared to a non-invasive control (placebo-IVt) without joint injection. The systemic cytokines/chemokines profile exhibited a time-dependent variation between transplant routes, displaying a transient anti-inflammatory systemic response for both MSCs-IVt and MSCs-IAt. A single injection of MSCs, whether administered via the intra-articular or intravenous route, performed 9 weeks after DMM surgery, did not effectively inhibit AC degeneration when compared to a non-invasive control.

3.
J Biol Chem ; 293(22): 8394-8409, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29618516

ABSTRACT

High-mobility group box 1 (HMGB1) is a chromatin-associated protein that, in response to stress or injury, translocates from the nucleus to the extracellular milieu, where it functions as an alarmin. HMGB1's function is in part determined by the complexes (HMGB1c) it forms with other molecules. However, structural modifications in the HMGB1 polypeptide that may regulate HMGB1c formation have not been previously described. In this report, we observed high-molecular weight, denaturing-resistant HMGB1c in the plasma and peripheral blood mononuclear cells of individuals with systemic lupus erythematosus (SLE) and, to a much lesser extent, in healthy subjects. Differential HMGB1c levels were also detected in mouse tissues and cultured cells, in which these complexes were induced by endotoxin or the immunological adjuvant alum. Of note, we found that HMGB1c formation is catalyzed by the protein-cross-linking enzyme transglutaminase-2 (TG2). Cross-link site mapping and MS analysis revealed that HMGB1 can be cross-linked to TG2 as well as a number of additional proteins, including human autoantigens. These findings have significant functional implications for studies of cellular stress responses and innate immunity in SLE and other autoimmune disease.


Subject(s)
Autoantigens/metabolism , GTP-Binding Proteins/metabolism , HMGB1 Protein/metabolism , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/metabolism , Transglutaminases/metabolism , Autoantigens/immunology , Cells, Cultured , GTP-Binding Proteins/immunology , HMGB1 Protein/immunology , Humans , Leukocytes, Mononuclear/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Molecular Weight , Protein Glutamine gamma Glutamyltransferase 2 , Substrate Specificity , Transglutaminases/immunology
4.
J Bone Miner Res ; 32(10): 2103-2115, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28585695

ABSTRACT

The Ras homolog A (RhoA) subfamily of Rho guanosine triphosphatases (GTPases) regulates actin-based cellular functions in bone such as differentiation, migration, and mechanotransduction. Polymorphisms or genetic ablation of RHOA and some of its regulatory guanine exchange factors (GEFs) have been linked to poor bone health in humans and mice, but the effects of RhoA-specific GTPase-activating proteins (GAPs) on bone quality have not yet been identified. Therefore, we examined the consequences of RhoGAP Myo9b gene knockout on bone growth, phenotype, and cellular activity. Male and female mice lacking both alleles demonstrated growth retardation and decreased bone formation rates during early puberty. These mice had smaller, weaker bones by 4 weeks of age, but only female KOs had altered cellular numbers, with fewer osteoblasts and more osteoclasts. By 12 weeks of age, bone quality in KOs worsened. In contrast, 4-week-old heterozygotes demonstrated bone defects that resolved by 12 weeks of age. Throughout, Myo9b ablation affected females more than males. Osteoclast activity appeared unaffected. In primary osteogenic cells, Myo9b was distributed in stress fibers and focal adhesions, and its absence resulted in poor spreading and eventual detachment from culture dishes. Similarly, MC3T3-E1 preosteoblasts with transiently suppressed Myo9b levels spread poorly and contained decreased numbers of focal adhesions. These cells also demonstrated reduced ability to undergo IGF-1-induced spreading or chemotaxis toward IGF-1, though responses to PDGF and BMP-2 were unaffected. IGF-1 receptor (IGF1R) activation was normal in cells with diminished Myo9b levels, but the activated receptor was redistributed from stress fibers and focal adhesions into nuclei, potentially affecting receptor accessibility and gene expression. These results demonstrate that Myo9b regulates a subset of RhoA-activated processes necessary for IGF-1 responsiveness in osteogenic cells, and is critical for normal bone formation in growing mice. © 2017 American Society for Bone and Mineral Research.


Subject(s)
Bone Development , Insulin-Like Growth Factor I/pharmacology , Myosins/metabolism , Osteoblasts/metabolism , Animals , Biomechanical Phenomena , Bone Development/drug effects , Cancellous Bone/metabolism , Cancellous Bone/pathology , Cancellous Bone/physiopathology , Cell Adhesion , Cell Line , Chemotaxis , Femur/metabolism , Femur/pathology , Femur/physiopathology , Gene Knockdown Techniques , Mice, Inbred C57BL , Mice, Knockout , Myosins/deficiency , Osteoblasts/drug effects , Rats , Sexual Maturation
5.
Front Physiol ; 8: 236, 2017.
Article in English | MEDLINE | ID: mdl-28491039

ABSTRACT

Daily moderate exercise (DME) and stress management are underemphasized in the care of patients with lupus nephritis (LN) due to a poor comprehensive understanding of their potential roles in controlling the inflammatory response. To investigate these effects on murine LN, disease progression was monitored with either DME or social disruption stress (SDR) induction in NZM2410/J mice, which spontaneously develop severe, early-onset LN. SDR of previously established social hierarchies was performed daily for 6 days and DME consisted of treadmill walking (8.5 m/min for 45 min/day). SDR significantly enhanced kidney disease when compared to age-matched, randomly selected control counterparts, as measured by histopathological analysis of H&E staining and immunohistochemistry for complement component 3 (C3) and IgG complex deposition. Conversely, while 88% of non-exercised mice displayed significant renal damage by 43 weeks of age, this was reduced to 45% with exercise. DME also reduced histopathology in kidney tissue and significantly decreased deposits of C3 and IgG complexes. Further examination of renal infiltrates revealed a macrophage-mediated inflammatory response that was significantly induced with SDR and suppressed with DME, which also correlated with expression of inflammatory mediators. Specifically, SDR induced IL-6, TNF-α, IL-1ß, and MCP-1, while DME suppressed IL-6, TNF-α, IL-10, CXCL1, and anti-dsDNA autoantibodies. These data demonstrate that psychological stressors and DME have significant, but opposing effects on the chronic inflammation associated with LN; thus identifying and characterizing stress reduction and a daily regimen of physical activity as potential adjunct therapies to complement pharmacological intervention in the management of autoimmune disorders, including LN.

6.
PLoS Negl Trop Dis ; 10(12): e0005220, 2016 12.
Article in English | MEDLINE | ID: mdl-28027305

ABSTRACT

BACKGROUND: Detection of Mycobacterium leprae in slit skin smear (SSS) is a gold standard technique for the leprosy diagnosis. Over recent years, molecular diagnosis by using PCR has been increasingly used as an alternative for its diagnosis due to its higher sensitivity. This study was carried out for comparative evaluation of PCR and SSS microscopy in a cohort of new leprosy cases diagnosed in B. P. Koirala Institute of health Sciences, Dharan, Nepal. METHODOLOGY/PRINCIPAL FINDINGS: In this prospective crossectional study, 50 new clinically diagnosed cases of leprosy were included. DNA was extracted from SSS and PCR was carried out to amplify 129 bp sequence of M. leprae repetitive element. Sensitivity of SSS and PCR was 18% and 72% respectively. Improvement of 54% case detection by PCR clearly showed its advantage over SSS. Furthermore, PCR could confirm the leprosy diagnosis in 66% of AFB negative cases indicating its superiority over SSS. In the paucibacillary (PB) patients, whose BI was zero; sensitivity of PCR was 44%, whereas it was 78% in the multibacillary patients. CONCLUSIONS/SIGNIFICANCE: Our study showed PCR to be more sensitive than SSS microscopy in diagnosing leprosy. Moreover, it explored the characteristic feature of PCR which detected higher level of early stage(PB) cases tested negative by SSS. Being an expensive technique, PCR may not be feasible in all the cases, however, it would be useful in diagnosis of early cases of leprosy as opposed to SSS.


Subject(s)
Leprosy/diagnosis , Mycobacterium leprae/isolation & purification , Polymerase Chain Reaction/methods , Skin/microbiology , Adolescent , Adult , Aged , Child , Cross-Sectional Studies , DNA, Bacterial/isolation & purification , Female , Humans , Male , Microscopy , Middle Aged , Nepal , Prospective Studies , ROC Curve , Sensitivity and Specificity , Young Adult
7.
J Orthop Res ; 33(2): 237-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25376614

ABSTRACT

To investigate how surgically created acute full-thickness cartilage defects of similar size and location created on the medial versus lateral femoral condyle influence progression of spontaneous cartilage lesions in a rat model. Full-thickness cartilage defects of 1 mm were surgically created on the medial or lateral femoral condyles on the right leg of 20 rats (n = 10/group). Ten rats served as controls. Spontaneous lesion progression on the ipsilateral and contralateral surfaces was examined using a high-resolution digital camera along with H&E and Safranin-O staining. Chondral defects were scored grossly and histologically. Control femur displayed no cartilage disruption. Surgically treated knees exhibited created and spontaneous cartilage defects with no evidence of healing unless subchondral bone was penetrated. Ipsilateral spontaneous lesions on the lateral condyle were significantly more severe on average (p = 0.009) compared to medial lesions on gross examination. Histological examination found contralateral lesions on the lateral surface following surgically created medial lesions to be more severe (p = 0.057) compared to contralateral lesions. A trend toward more susceptible chondral damage to the lateral condyle was observed following acute lesion creation on either medial or lateral condyles. Mechanisms behind this pattern of spontaneous lesion development are unclear, requring further investigation.


Subject(s)
Disease Progression , Knee Joint/pathology , Osteoarthritis, Knee/pathology , Animals , Disease Models, Animal , Female , Random Allocation , Rats, Sprague-Dawley
8.
Ann Biomed Eng ; 42(1): 36-49, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23943071

ABSTRACT

Massage therapy has a long history and has been widely believed effective in restoring tissue function, relieving pain and stress, and promoting overall well-being. However, the application of massage-like actions and the efficacy of massage are largely based on anecdotal experiences that are difficult to define and measure. This leads to a somewhat limited evidence-based interface of massage therapy with modern medicine. In this study, we introduce a mechatronic device that delivers highly reproducible massage-like mechanical loads to the hind limbs of small animals (rats and rabbits), where various massage-like actions are quantified by the loading parameters (magnitude, frequency and duration) of the compressive and transverse forces on the subject tissues. The effect of massage is measured by the difference in passive viscoelastic properties of the subject tissues before and after mechanical loading, both obtained by the same device. Results show that this device is useful in identifying the loading parameters that are most conducive to a change in tissue mechanical properties, and can determine the range of loading parameters that result in sustained changes in tissue mechanical properties and function. This device presents the first step in our effort for quantifying the application of massage-like actions used clinically and measurement of their efficacy that can readily be combined with various quantitative measures (e.g., active mechanical properties and physiological assays) for determining the therapeutic and mechanistic effects of massage therapies.


Subject(s)
Massage/instrumentation , Animals , Female , Massage/methods , Models, Biological , Rabbits , Rats , Rats, Sprague-Dawley
9.
Antioxid Redox Signal ; 20(6): 970-85, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-23815527

ABSTRACT

SIGNIFICANCE: Mechanosignaling is vital for maintaining the structural integrity of bone under physiologic conditions. These signals activate and suppress multiple signaling cascades regulating bone formation and resorption. Understanding these pathways is of prime importance to exploit their therapeutic potential in disorders associated with bone loss due to disuse, trauma, or disruption of homeostatic mechanisms. RECENT ADVANCES: In the case of cells of the bone, an impressive amount of data has been generated that provides evidence of a complex mechanism by which mechanical signals can maintain or disrupt cellular homeostasis by driving transcriptional regulation of growth factors, matrix proteins and inflammatory mediators in health and inflammation. Mechanical signals act on cells in a magnitude dependent manner to induce bone deposition or resorption. During health, physiological levels of these signals are essential for maintaining bone strength and architecture, whereas during inflammation, similar signals can curb inflammation by suppressing the nuclear factor kappa B (NF-κB) signaling cascade, while upregulating matrix synthesis via mothers against decapentaplegic homolog and/or Wnt signaling cascades. Contrarily, excessive mechanical forces can induce inflammation via activation of the NF-κB signaling cascade. CRITICAL ISSUES: Given the osteogenic potential of mechanical signals, it is imperative to exploit their therapeutic efficacy for the treatment of bone disorders. Here we review select signaling pathways and mediators stimulated by mechanical signals to modulate the strength and integrity of the bone. FUTURE DIRECTIONS: Understanding the mechanisms of mechanotransduction and its effects on bone lay the groundwork for development of nonpharmacologic mechanostimulatory approaches for osteodegenerative diseases and optimal bone health.


Subject(s)
Inflammation/metabolism , Mechanotransduction, Cellular/physiology , Wounds and Injuries/metabolism , Animals , Bone and Bones/metabolism , Humans , NF-kappa B/metabolism
10.
Arthroscopy ; 29(10): 1722-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23890952

ABSTRACT

PURPOSE: To determine whether the basic science evidence supports the use of continuous passive motion (CPM) after articular cartilage injury in the knee. METHODS: A systematic review was performed identifying and evaluating studies in animal models that focused on the basic science of CPM of the knee. Databases included in this review were PubMed, Biosis Previews, SPORTDiscus, PEDro, and EMBASE. All functional, gross anatomic, histologic, and histochemical outcomes were extracted and analyzed. RESULTS: Primary outcomes of CPM analyzed in rabbit animal models (19 studies) included histologic changes in articular cartilage (13 studies), biomechanical changes and nutrition of intra-articular tissue (3 studies), and anti-inflammatory biochemical changes (3 studies). Nine studies specifically examined osteochondral defects, 6 of which used autogenous periosteal grafts. Other pathologies included were antigen-induced arthritis, septic arthritis, medial collateral ligament reconstruction, hemarthrosis, and chymopapain-induced proteoglycan destruction. In comparison to immobilized knees, CPM therapy led to decreased joint stiffness and complications related to adhesions while promoting improved neochondrogenesis with formation and preservation of normal articular cartilage. CPM was also shown to create a strong anti-inflammatory environment by effectively clearing harmful, inflammatory particles from within the knee. CONCLUSIONS: Current basic science evidence from rabbit studies has shown that CPM for the knee significantly improves motion and biological properties of articular cartilage. This may be translated to potentially improved outcomes in the management of articular cartilage pathology of the knee. CLINICAL RELEVANCE: If the rabbit model is relevant to humans, CPM may contribute to improved knee health by preventing joint stiffness, preserving normal articular tissue with better histologic and biologic properties, and improving range of motion as compared with joint immobilization and intermittent active motion.


Subject(s)
Cartilage, Articular/injuries , Joint Diseases/therapy , Motion Therapy, Continuous Passive , Range of Motion, Articular/physiology , Animals , Arthritis/physiopathology , Arthritis/therapy , Biomechanical Phenomena/physiology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/physiopathology , Joint Diseases/metabolism , Joint Diseases/physiopathology , Knee Joint , Models, Animal , Rabbits , Recovery of Function/physiology
11.
J Clin Periodontol ; 40(4): 387-95, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23432761

ABSTRACT

AIM: To investigate and compare outcomes following alveolar ridge preservation (ARP) in posterior maxilla and mandible. METHODS: Twenty-four patients (54 ± 3 years) with single posterior tooth extraction were included. ARP was performed with freeze-dried bone allograft and collagen membrane. Clinical parameters were recorded at extraction and re-entry. Harvested bone cores were analysed by microcomputed tomography (micro-CT), histomorphometry and immunohistochemistry. RESULTS: In both jaws, ARP prevented ridge height loss, but ridge width was significantly reduced by approximately 2.5 mm. Healing time, initial clinical attachment loss and amount of keratinized tissue at extraction site were identified as determinants of ridge height outcome. Buccal plate thickness and tooth root length were identified as determinants of ridge width outcome. In addition, initial ridge width was positively correlated with ridge width loss. Micro-CT revealed greater mineralization per unit volume in new bone compared with existing bone in mandible (p < 0.001). Distributions of residual graft, new cellular bone and immature tissue were similar in both jaws. CONCLUSION: Within the limitations of this study, the results indicate that in different anatomic locations different factors may determine ARP outcomes. Further studies are needed to better understand determinants of ARP outcomes.


Subject(s)
Alveolar Bone Loss/prevention & control , Alveolar Process/anatomy & histology , Bone Regeneration , Dental Implantation, Endosseous/methods , Guided Tissue Regeneration, Periodontal/methods , Tooth Socket/surgery , Adult , Aged , Aged, 80 and over , Alveolar Bone Loss/etiology , Alveolar Process/diagnostic imaging , Bone Transplantation , Chi-Square Distribution , Collagen/therapeutic use , Female , Humans , Male , Membranes, Artificial , Middle Aged , Prospective Studies , Regression Analysis , Statistics, Nonparametric , Tooth Extraction/adverse effects , Tooth Extraction/methods , Treatment Outcome , X-Ray Microtomography , Young Adult
12.
Tissue Eng Part A ; 19(5-6): 783-92, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23198877

ABSTRACT

Osteochondral tissue-engineered grafts are proposed to hold greater potential to repair/regenerate damaged cartilage through enhanced biochemical and mechanical interactions with underlying subchondral bone as compared to simple engineered cartilage. Additionally, biomechanical stimulation of articular chondrocytes (ACs) or osteoblasts (OBs) was shown to induce greater morphogenesis of the engineered tissues composed of these cells. In this report, to define the advantages of biomechanical stimulation to osteochondral grafts for tissue engineering, we examined whether (1) ACs and OBs in three-dimensional (3D) osteochondral constructs support functional development of each other at the molecular level, and (2) biomechanical stimulation of osteochondral constructs further promotes the regenerative potential of such grafts. Various configurations of cell/scaffold assemblies, including chondral, osseous, and osteochondral constructs, were engineered with mechano-responsive electrospun poly(ɛ-caprolactone) scaffolds. These constructs were subjected to either static or dynamic (10% cyclic compressive strain at 1 Hz for 3 h/day) culture conditions for 2 weeks. The expression of bone morphogenetic proteins (BMPs) was examined to assess the regenerative potential of each treatment on the cells. Biomechanical stimulation augmented a marked upregulation of Bmp2, Bmp6, and Bmp7 as well as downregulation of BMP antagonist, Bmp3, in a time-specific manner in the ACs and OBs of 3D osteochondral constructs. More importantly, the presence of biomechanically stimulated OBs was especially crucial for the induction of Bmp6 in ACs, a BMP required for chondrocytic growth and differentiation. Biomechanical stimulation led to enhanced tissue morphogenesis possibly through this BMP regulation, evident by the improved effective compressive modulus of the osteochondral constructs (710 kPa of dynamic culture vs. 280 kPa of static culture). Similar BMP regulation was observed in the femoral cartilages of the rats subjected to gentle exercise, demonstrating the physiological relevance of in vitro biomechanical stimulation of osteochondral constructs. Overall, our findings show that biomechanical stimulation may be critical for cross signaling between ACs and OBs to support chondrocytic growth in 3D osteochondral tissues.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Chondrocytes/cytology , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Biomechanical Phenomena/drug effects , Bone Morphogenetic Proteins/pharmacology , Cartilage, Articular/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Compressive Strength/drug effects , Female , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Paracrine Communication/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors
13.
PLoS One ; 7(6): e39365, 2012.
Article in English | MEDLINE | ID: mdl-22724005

ABSTRACT

BACKGROUND: Osteoporosis is a bone disorder associated with loss of bone mineral density and micro architecture. A balance of osteoblasts and osteoclasts activities maintains bone homeostasis. Increased bone loss due to increased osteoclast and decreased osteoblast activities is considered as an underlying cause of osteoporosis. METHODS AND FINDINGS: The cures for osteoporosis are limited, consequently the potential of CD34+ cell therapies is currently being considered. We developed a nanofiber-based expansion technology to obtain adequate numbers of CD34(+) cells isolated from human umbilical cord blood, for therapeutic applications. Herein, we show that CD34(+) cells could be differentiated into osteoblastic lineage, in vitro. Systemically delivered CD34(+) cells home to the bone marrow and significantly improve bone deposition, bone mineral density and bone micro-architecture in osteoporotic mice. The elevated levels of osteocalcin, IL-10, GM-CSF, and decreased levels of MCP-1 in serum parallel the improvements in bone micro-architecture. Furthermore, CD34(+) cells improved osteoblast activity and concurrently impaired osteoclast differentiation, maturation and functionality. CONCLUSIONS: These findings demonstrate a novel approach utilizing nanofiber-expanded CD34(+) cells as a therapeutic application for the treatment of osteoporosis.


Subject(s)
Antigens, CD34 , Cord Blood Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Osteoblasts/cytology , Osteoclasts/cytology , Osteoporosis/therapy , Animals , Antigens, CD34/metabolism , Bone Marrow/metabolism , Bone and Bones/ultrastructure , Calcification, Physiologic , Cell Culture Techniques , Cell Differentiation , Cytokines/blood , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Osteoblasts/metabolism , Osteocalcin/blood , Osteoclasts/metabolism , Osteogenesis , Osteoporosis/metabolism
14.
PLoS One ; 6(9): e24320, 2011.
Article in English | MEDLINE | ID: mdl-21931681

ABSTRACT

Chronic inflammation is one of the major causes of cartilage destruction in osteoarthritis. Here, we systematically analyzed the changes in gene expression associated with the progression of cartilage destruction in monoiodoacetate-induced arthritis (MIA) of the rat knee. Sprague Dawley female rats were given intra-articular injection of monoiodoacetate in the knee. The progression of MIA was monitored macroscopically, microscopically and by micro-computed tomography. Grade 1 damage was observed by day 5 post-monoiodoacetate injection, progressively increasing to Grade 2 by day 9, and to Grade 3-3.5 by day 21. Affymetrix GeneChip was utilized to analyze the transcriptome-wide changes in gene expression, and the expression of salient genes was confirmed by real-time-PCR. Functional networks generated by Ingenuity Pathways Analysis (IPA) from the microarray data correlated the macroscopic/histologic findings with molecular interactions of genes/gene products. Temporal changes in gene expression during the progression of MIA were categorized into five major gene clusters. IPA revealed that Grade 1 damage was associated with upregulation of acute/innate inflammatory responsive genes (Cluster I) and suppression of genes associated with musculoskeletal development and function (Cluster IV). Grade 2 damage was associated with upregulation of chronic inflammatory and immune trafficking genes (Cluster II) and downregulation of genes associated with musculoskeletal disorders (Cluster IV). The Grade 3 to 3.5 cartilage damage was associated with chronic inflammatory and immune adaptation genes (Cluster III). These findings suggest that temporal regulation of discrete gene clusters involving inflammatory mediators, receptors, and proteases may control the progression of cartilage destruction. In this process, IL-1ß, TNF-α, IL-15, IL-12, chemokines, and NF-κB act as central nodes of the inflammatory networks, regulating catabolic processes. Simultaneously, upregulation of asporin, and downregulation of TGF-ß complex, SOX-9, IGF and CTGF may be central to suppress matrix synthesis and chondrocytic anabolic activities, collectively contributing to the progression of cartilage destruction in MIA.


Subject(s)
Arthritis/genetics , Arthritis/pathology , Disease Progression , Gene Expression Regulation , Animals , Arthritis/chemically induced , Arthritis/diagnostic imaging , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Proliferation , Cluster Analysis , Extracellular Matrix/metabolism , Female , Femur/diagnostic imaging , Femur/pathology , Gene Regulatory Networks/genetics , Immunity, Innate/genetics , Inflammation/complications , Inflammation/genetics , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Iodoacetic Acid , Joints/pathology , Multigene Family/genetics , Oligonucleotide Array Sequence Analysis , Radiography , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Transcriptome , Up-Regulation/genetics
15.
Arthritis Rheum ; 63(6): 1613-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21400474

ABSTRACT

OBJECTIVE: Physiotherapies are the most widely recommended conservative treatment for arthritic diseases. The present study was undertaken to examine the molecular mechanisms underlying the effects of gentle treadmill walking (GTW) on various stages of monoiodoacetate-induced arthritis (MIA) to elucidate the basis for the success or failure of such therapies in joint damage. METHODS: Knees were obtained from untreated control rats, rats with MIA that did not undergo GTW, rats with MIA in which GTW regimens were started 1 day post-MIA induction, and rats with MIA in which GTW regimens were started after cartilage damage had progressed to grade 1 or grade 2. The cartilage was examined macroscopically, microscopically, and by microfocal computed tomography imaging. Transcriptome-wide gene expression analysis was performed, and microarray data were assessed by Ingenuity Pathways Analysis to identify molecular functional networks regulated by GTW. RESULTS: GTW intervention started on day 1 post-MIA induction significantly prevented the progression of MIA, but its efficacy was reduced when implemented on knees exhibiting close to grade 1 cartilage damage. GTW accelerated cartilage damage in knees with close to grade 2 damage. Transcriptome-wide gene expression analysis revealed that GTW intervention started 1 day post-MIA inception significantly suppressed inflammation-associated genes and up-regulated matrix-associated gene networks. However, delayed GTW intervention after grade 1 damage had occurred was less effective in suppressing proinflammatory genes or up-regulating matrix synthesis. CONCLUSION: The present findings suggest that GTW suppresses proinflammatory gene networks and up-regulates matrix synthesis to prevent progression of cartilage damage in MIA-affected knees. However, the extent of cartilage damage at the initiation of GTW may be an important determinant of the success or failure of such therapies.


Subject(s)
Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Exercise Therapy , Walking , Animals , Arthritis, Experimental/chemically induced , Cartilage/metabolism , Cartilage/pathology , Disease Progression , Exercise Test , Female , Gene Expression Profiling , Iodoacetic Acid/pharmacology , Knee Joint/pathology , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Up-Regulation
16.
PLoS One ; 6(3): e17161, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21390242

ABSTRACT

BACKGROUND: Mice deficient in the large zinc finger protein, ZAS3, show postnatal increase in bone mass suggesting that ZAS3 is critical in the regulation of bone homeostasis. Although ZAS3 has been shown to inhibit osteoblast differentiation, its role on osteoclastogenesis has not been determined. In this report we demonstrated the role of ZAS3 in bone resorption by examining the signaling mechanisms involved in osteoclastogenesis. METHODOLOGY/PRINCIPAL FINDINGS: Comparison of adult wild-type and ZAS3 knockout (ZAS3-/-) mice showed that ZAS3 deficiency led to thicker bones that are more resistant to mechanical fracture. Additionally, ZAS3-/- bones showed fewer osteoclasts and inefficient M-CSF/sRANKL-mediated osteoclastogenesis ex vivo. Utilizing RAW 264.7 pre-osteoclasts, we demonstrated that overexpression of ZAS3 promoted osteoclastogenesis and the expression of crucial osteoclastic molecules, including phospho-p38, c-Jun, NFATc1, TRAP and CTSK. Contrarily, ZAS3 silencing by siRNA inhibited osteoclastogenesis. Co-immunoprecipitation experiments demonstrated that ZAS3 associated with TRAF6, the major receptor associated molecule in RANK signaling. Furthermore, EMSA suggested that nuclear ZAS3 could regulate transcription by binding to gene regulatory elements. CONCLUSION/SIGNIFICANCE: Collectively, the data suggested a novel role of ZAS3 as a positive regulator of osteoclast differentiation. ZAS3 deficiency caused increased bone mass, at least in part due to decreased osteoclast formation and bone resorption. These functions of ZAS3 were mediated via activation of multiple intracellular targets. In the cytoplasmic compartment, ZAS3 associated with TRAF6 to control NF-kB and MAP kinase signaling cascades. Nuclear ZAS3 acted as a transcriptional regulator for osteoclast-associated genes. Additionally, ZAS3 activated NFATc1 required for the integration of RANK signaling in the terminal differentiation of osteoclasts. Thus, ZAS3 was a crucial molecule in osteoclast differentiation, which might potentially serve as a target in the design of therapeutic interventions for the treatment of bone diseases related to increased osteoclast activity such as postmenopausal osteoporosis, Paget's disease, and rheumatoid arthritis.


Subject(s)
DNA-Binding Proteins/metabolism , Osteoclasts/metabolism , Osteogenesis , Transcription Factors/metabolism , Zinc Fingers , Aging/drug effects , Aging/metabolism , Amino Acid Sequence , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Count , Cell Line , DNA-Binding Proteins/deficiency , Femur/drug effects , Femur/metabolism , Femur/pathology , Fractures, Bone/pathology , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Mice , Models, Biological , Molecular Sequence Data , Osteoclasts/drug effects , Osteoclasts/pathology , Osteogenesis/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , RANK Ligand/pharmacology , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , TNF Receptor-Associated Factor 6/chemistry , TNF Receptor-Associated Factor 6/metabolism , Transcription Factors/deficiency
17.
Acta Biomater ; 7(4): 1516-24, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21109030

ABSTRACT

As the potential range of stem cell applications in tissue engineering continues to grow, the appropriate scaffolding choice is necessary to create tightly defined artificial microenvironments for each target organ. These microenvironments determine stem cell fate via control over differentiation. In this study we examined the specific effects of scaffold stiffness on embryonic mesenchymal progenitor cell behavior. Mechanically distinct scaffolds having identical microstructures and surface chemistries were produced utilizing core-shell electrospinning. The modulus of core-shell poly(ether sulfone)-poly(ε-caprolactone) (PES-PCL) fibers (30.6 MPa) was more than four times that of pure PCL (7.1 MPa). The results for chondrogenic and osteogenic differentiation of progenitor cells on each scaffold indicate that the lower modulus PCL fibers provided more appropriate microenvironments for chondrogenesis, evident by a marked up-regulation of chondrocytic Sox9, collagen type 2, and aggrecan gene expression and chondrocyte-specific extracellular matrix glycosaminoglycan production. In contrast, the stiffer core-shell PES-PCL fibers supported enhanced osteogenesis by promoting osteogenic Runx2, alkaline phosphatase, and osteocalcin gene expression, as well as alkaline phosphatase activity. The findings demonstrate that the microstructural stiffness/modules of a scaffold and the pliability of individual fibers may play a critical role in controlling stem cell differentiation. Regulation of cytoskeletal organization may occur via a "dynamic scaffold" leading to the subsequent intracellular signaling events that control differentiation-specific gene expression.


Subject(s)
Cell Differentiation , Elastic Modulus , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Nanofibers/chemistry , Animals , Cell Differentiation/drug effects , Cell Line , Chondrogenesis/drug effects , Chondrogenesis/genetics , Elastic Modulus/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Nanofibers/ultrastructure , Osteogenesis/drug effects , Osteogenesis/genetics , Photoelectron Spectroscopy , Polyesters/pharmacology , Polymers/pharmacology , Sulfones/pharmacology , Tissue Engineering/methods , Tissue Scaffolds/chemistry
18.
Arthritis Res Ther ; 12(3): R106, 2010.
Article in English | MEDLINE | ID: mdl-20509944

ABSTRACT

INTRODUCTION: The importance of mechanical signals in normal and inflamed cartilage is well established. Chondrocytes respond to changes in the levels of proinflammatory cytokines and mechanical signals during inflammation. Cytokines like interleukin (IL)-1beta suppress homeostatic mechanisms and inhibit cartilage repair and cell proliferation. However, matrix synthesis and chondrocyte (AC) proliferation are upregulated by the physiological levels of mechanical forces. In this study, we investigated intracellular mechanisms underlying reparative actions of mechanical signals during inflammation. METHODS: ACs isolated from articular cartilage were exposed to low/physiologic levels of dynamic strain in the presence of IL-1beta. The cell extracts were probed for differential activation/inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling cascade. The regulation of gene transcription was examined by real-time polymerase chain reaction. RESULTS: Mechanoactivation, but not IL-1beta treatment, of ACs initiated integrin-linked kinase activation. Mechanical signals induced activation and subsequent C-Raf-mediated activation of MAP kinases (MEK1/2). However, IL-1beta activated B-Raf kinase activity. Dynamic strain did not induce B-Raf activation but instead inhibited IL-1beta-induced B-Raf activation. Both mechanical signals and IL-1beta induced ERK1/2 phosphorylation but discrete gene expression. ERK1/2 activation by mechanical forces induced SRY-related protein-9 (SOX-9), vascular endothelial cell growth factor (VEGF), and c-Myc mRNA expression and AC proliferation. However, IL-1beta did not induce SOX-9, VEGF, and c-Myc gene expression and inhibited AC cell proliferation. More importantly, SOX-9, VEGF, and Myc gene transcription and AC proliferation induced by mechanical signals were sustained in the presence of IL-1beta. CONCLUSIONS: The findings suggest that mechanical signals may sustain their effects in proinflammatory environments by regulating key molecules in the MAP kinase signaling cascade. Furthermore, the findings point to the potential of mechanosignaling in cartilage repair during inflammation.


Subject(s)
Chondrocytes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-myc/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Biomechanical Phenomena , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Chondrocytes/cytology , Female , Inflammation/metabolism , Interleukin-1beta/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Animal , Rats , Rats, Sprague-Dawley
19.
J Immunol ; 185(2): 1215-21, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20548028

ABSTRACT

Signals generated by the dynamic mechanical strain critically regulate endothelial cell proliferation and angiogenesis; however, the molecular basis remains unclear. We investigated the mechanisms by which human dermal microvascular endothelial cells (HDMECs) perceive mechanical signals and relay them intracellularly to regulate gene expression and endothelial cell proliferation. HDMECs were exposed to low/physiologic levels of dynamic strain and probed for the differential activation/inhibition of kinases in the mechanosignaling cascade associated with endothelial cell gene activation. Because angiogenesis is important at inflammatory sites, we also assessed the mechanisms of mechanosignaling in the presence of an proinflammatory cytokine IL-1beta. In this article, we demonstrate that the mechanosignaling cascade is initiated by vascular endothelial growth receptor-2 (VEGFR2) activation. Mechanoactivation of VEGFR2 results in its nuclear translocation and elevation of PI3K-dependent Ser473-Akt phosphorylation. Subsequently, activated Akt inactivates the kinase activity of the serine/threonine kinase, glycogen synthase kinase-3beta (GSK3beta), via its Ser9 phosphorylation. Thus, inactive GSK3beta fails to phosphorylate cyclin D1 and prevents its proteosomal degradation and, consequently, promotes endothelial cell survival and proliferation. In the presence of IL-1beta, cyclin D1 is phosphorylated and degraded, leading to inhibition of cell proliferation. However, mechanical signals repress cyclin D1 phosphorylation and upregulate cell proliferation, despite the presence of IL-1beta. The data indicate that the VEGFR2/Akt/GSK3beta signaling cascade plays a critical role in sensing and phospho-relaying mechanical stimuli in endothelial cells. Furthermore, mechanical forces control highly interconnected networks of proinflammatory and Akt signaling cascades to upregulate endothelial cell proliferation.


Subject(s)
Cell Proliferation , Endothelial Cells/immunology , Signal Transduction/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cyclin D1/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Stress, Mechanical , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
PLoS One ; 4(4): e5262, 2009.
Article in English | MEDLINE | ID: mdl-19370157

ABSTRACT

BACKGROUND: During normal physical activities cartilage experiences dynamic compressive forces that are essential to maintain cartilage integrity. However, at non-physiologic levels these signals can induce inflammation and initiate cartilage destruction. Here, by examining the pro-inflammatory signaling networks, we developed a mathematical model to show the magnitude-dependent regulation of chondrocytic responses by compressive forces. METHODOLOGY/PRINCIPAL FINDINGS: Chondrocytic cells grown in 3-D scaffolds were subjected to various magnitudes of dynamic compressive strain (DCS), and the regulation of pro-inflammatory gene expression via activation of nuclear factor-kappa B (NF-kappaB) signaling cascade examined. Experimental evidences provide the existence of a threshold in the magnitude of DCS that regulates the mRNA expression of nitric oxide synthase (NOS2), an inducible pro-inflammatory enzyme. Interestingly, below this threshold, DCS inhibits the interleukin-1beta (IL-1beta)-induced pro-inflammatory gene expression, with the degree of suppression depending on the magnitude of DCS. This suppression of NOS2 by DCS correlates with the attenuation of the NF-kappaB signaling pathway as measured by IL-1beta-induced phosphorylation of the inhibitor of kappa B (IkappaB)-alpha, degradation of IkappaB-alpha and IkappaB-beta, and subsequent nuclear translocation of NF-kappaB p65. A mathematical model developed to understand the complex dynamics of the system predicts two thresholds in the magnitudes of DCS, one for the inhibition of IL-1beta-induced expression of NOS2 by DCS at low magnitudes, and second for the DCS-induced expression of NOS2 at higher magnitudes. CONCLUSIONS/SIGNIFICANCE: Experimental and computational results indicate that biomechanical signals suppress and induce inflammation at critical thresholds through activation/suppression of the NF-kappaB signaling pathway. These thresholds arise due to the bistable behavior of the networks originating from the positive feedback loop between NF-kappaB and its target genes. These findings lay initial groundwork for the identification of the thresholds in physical activities that can differentiate its favorable actions from its unfavorable consequences on joints.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Inflammation/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Stress, Mechanical , Biomechanical Phenomena , Cartilage/cytology , Cartilage/physiopathology , Cell Line , Gene Expression Regulation , Humans , I-kappa B Proteins/metabolism , Inflammation/genetics , Inflammation/physiopathology , Interleukin-1beta/metabolism , Models, Biological , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Nitric Oxide Synthase/genetics , Protein Transport , RNA, Messenger/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...